Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
3.
Nature ; 627(8004): 671-679, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38448585

ABSTRACT

DNA and histone modifications combine into characteristic patterns that demarcate functional regions of the genome1,2. While many 'readers' of individual modifications have been described3-5, how chromatin states comprising composite modification signatures, histone variants and internucleosomal linker DNA are interpreted is a major open question. Here we use a multidimensional proteomics strategy to systematically examine the interaction of around 2,000 nuclear proteins with over 80 modified dinucleosomes representing promoter, enhancer and heterochromatin states. By deconvoluting complex nucleosome-binding profiles into networks of co-regulated proteins and distinct nucleosomal features driving protein recruitment or exclusion, we show comprehensively how chromatin states are decoded by chromatin readers. We find highly distinctive binding responses to different features, many factors that recognize multiple features, and that nucleosomal modifications and linker DNA operate largely independently in regulating protein binding to chromatin. Our online resource, the Modification Atlas of Regulation by Chromatin States (MARCS), provides in-depth analysis tools to engage with our results and advance the discovery of fundamental principles of genome regulation by chromatin states.


Subject(s)
Chromatin Assembly and Disassembly , Chromatin , Nuclear Proteins , Nucleosomes , Proteomics , Humans , Binding Sites , Chromatin/chemistry , Chromatin/genetics , Chromatin/metabolism , DNA/genetics , DNA/metabolism , Enhancer Elements, Genetic , Heterochromatin/genetics , Heterochromatin/metabolism , Histones/metabolism , Nuclear Proteins/analysis , Nuclear Proteins/metabolism , Nucleosomes/chemistry , Nucleosomes/genetics , Nucleosomes/metabolism , Promoter Regions, Genetic , Protein Binding , Proteomics/methods
4.
Nat Cell Biol ; 25(7): 1017-1032, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37414849

ABSTRACT

Chromatin is dynamically reorganized when DNA replication forks are challenged. However, the process of epigenetic reorganization and its implication for fork stability is poorly understood. Here we discover a checkpoint-regulated cascade of chromatin signalling that activates the histone methyltransferase EHMT2/G9a to catalyse heterochromatin assembly at stressed replication forks. Using biochemical and single molecule chromatin fibre approaches, we show that G9a together with SUV39h1 induces chromatin compaction by accumulating the repressive modifications, H3K9me1/me2/me3, in the vicinity of stressed replication forks. This closed conformation is also favoured by the G9a-dependent exclusion of the H3K9-demethylase JMJD1A/KDM3A, which facilitates heterochromatin disassembly upon fork restart. Untimely heterochromatin disassembly from stressed forks by KDM3A enables PRIMPOL access, triggering single-stranded DNA gap formation and sensitizing cells towards chemotherapeutic drugs. These findings may help in explaining chemotherapy resistance and poor prognosis observed in patients with cancer displaying elevated levels of G9a/H3K9me3.


Subject(s)
Heterochromatin , Histones , Humans , Histones/genetics , Histones/metabolism , Heterochromatin/genetics , Chromatin/genetics , Chromatin Assembly and Disassembly , DNA Replication , Histocompatibility Antigens , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics
5.
Cell Rep ; 42(2): 112045, 2023 02 28.
Article in English | MEDLINE | ID: mdl-36701236

ABSTRACT

The chromatin environment at origins of replication is thought to influence DNA replication initiation in eukaryotic genomes. However, it remains unclear how and which chromatin features control the firing of early-efficient (EE) or late-inefficient (LI) origins. Here, we use site-specific recombination and single-locus chromatin isolation to purify EE and LI replication origins in Saccharomyces cerevisiae. Using mass spectrometry, we define the protein composition of native chromatin regions surrounding the EE and LI replication start sites. In addition to known origin interactors, we find the microtubule-binding Ask1/DASH complex as an origin-regulating factor. Strikingly, tethering of Ask1 to individual origin sites advances replication timing (RT) of the targeted chromosomal domain. Targeted degradation of Ask1 globally changes RT of a subset of origins, which can be reproduced by inhibiting microtubule dynamics. Thus, our findings mechanistically connect RT and chromosomal organization via Ask1/DASH with the microtubule cytoskeleton.


Subject(s)
Microtubule-Associated Proteins , Replication Origin , Saccharomyces cerevisiae Proteins , Chromatin/metabolism , DNA/metabolism , DNA Replication , DNA Replication Timing , Microtubule-Associated Proteins/metabolism , Multiprotein Complexes/metabolism , Proteomics , Replication Origin/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism
6.
Nucleic Acids Res ; 50(17): 9705-9723, 2022 09 23.
Article in English | MEDLINE | ID: mdl-36095123

ABSTRACT

Trypanosomes diverged from the main eukaryotic lineage about 600 million years ago, and display some unusual genomic and epigenetic properties that provide valuable insight into the early processes employed by eukaryotic ancestors to regulate chromatin-mediated functions. We analysed Trypanosoma brucei core histones by high mass accuracy middle-down mass spectrometry to map core histone post-translational modifications (PTMs) and elucidate cis-histone combinatorial PTMs (cPTMs). T. brucei histones are heavily modified and display intricate cPTMs patterns, with numerous hypermodified cPTMs that could contribute to the formation of non-repressive euchromatic states. The Trypanosoma brucei H2A C-terminal tail is hyperacetylated, containing up to five acetylated lysine residues. MNase-ChIP-seq revealed a striking enrichment of hyperacetylated H2A at Pol II transcription start regions, and showed that H2A histones that are hyperacetylated in different combinations localised to different genomic regions, suggesting distinct epigenetic functions. Our genomics and proteomics data provide insight into the complex epigenetic mechanisms used by this parasite to regulate a genome that lacks the transcriptional control mechanisms found in later-branched eukaryotes. The findings further demonstrate the complexity of epigenetic mechanisms that were probably shared with the last eukaryotic common ancestor.


Subject(s)
Histones/metabolism , Protozoan Proteins/metabolism , Trypanosoma brucei brucei , Chromatin/genetics , Histone Code , Lysine/metabolism , Protein Processing, Post-Translational , Trypanosoma brucei brucei/genetics , Trypanosoma brucei brucei/metabolism
7.
Nucleic Acids Res ; 50(15): 8491-8511, 2022 08 26.
Article in English | MEDLINE | ID: mdl-35904814

ABSTRACT

DNA methylation (5-methylcytosine (5mC)) is critical for genome stability and transcriptional regulation in mammals. The discovery that ten-eleven translocation (TET) proteins catalyze the oxidation of 5mC to 5-hydroxymethylcytosine (5hmC), 5-formylcytosine (5fC), and 5-carboxylcytosine (5caC) revolutionized our perspective on the complexity and regulation of DNA modifications. However, to what extent the regulatory functions of TET1 can be attributed to its catalytic activity remains unclear. Here, we use genome engineering and quantitative multi-omics approaches to dissect the precise catalytic vs. non-catalytic functions of TET1 in murine embryonic stem cells (mESCs). Our study identifies TET1 as an essential interaction hub for multiple chromatin modifying complexes and a global regulator of histone modifications. Strikingly, we find that the majority of transcriptional regulation depends on non-catalytic functions of TET1. In particular, we show that TET1 is critical for the establishment of H3K9me3 and H4K20me3 at endogenous retroviral elements (ERVs) and their silencing that is independent of its canonical role in DNA demethylation. Furthermore, we provide evidence that this repression of ERVs depends on the interaction between TET1 and SIN3A. In summary, we demonstrate that the non-catalytic functions of TET1 are critical for regulation of gene expression and the silencing of endogenous retroviruses in mESCs.


Subject(s)
DNA-Binding Proteins/metabolism , Endogenous Retroviruses , Proto-Oncogene Proteins/metabolism , 5-Methylcytosine/metabolism , Animals , Cytosine/metabolism , DNA Demethylation , DNA Methylation , DNA-Binding Proteins/genetics , Endogenous Retroviruses/genetics , Endogenous Retroviruses/metabolism , Gene Expression , Mammals/genetics , Mice , Proto-Oncogene Proteins/genetics
8.
Life Sci Alliance ; 5(11)2022 11.
Article in English | MEDLINE | ID: mdl-35820704

ABSTRACT

The mitotic deacetylase complex MiDAC has recently been shown to play a vital physiological role in embryonic development and neurite outgrowth. However, how MiDAC functionally intersects with other chromatin-modifying regulators is poorly understood. Here, we describe a physical interaction between the histone H3K27 demethylase UTX, a complex-specific subunit of the enhancer-associated MLL3/4 complexes, and MiDAC. We demonstrate that UTX bridges the association of the MLL3/4 complexes and MiDAC by interacting with ELMSAN1, a scaffolding subunit of MiDAC. Our data suggest that MiDAC constitutes a negative genome-wide regulator of H4K20ac, an activity which is counteracted by the MLL3/4 complexes. MiDAC and the MLL3/4 complexes co-localize at many genomic regions, which are enriched for H4K20ac and the enhancer marks H3K4me1, H3K4me2, and H3K27ac. We find that MiDAC antagonizes the recruitment of UTX and MLL4 and negatively regulates H4K20ac, and to a lesser extent H3K4me2 and H3K27ac, resulting in transcriptional attenuation of associated genes. In summary, our findings provide a paradigm how the opposing roles of chromatin-modifying components, such as MiDAC and the MLL3/4 complexes, balance the transcriptional output of specific gene expression programs.


Subject(s)
Enhancer Elements, Genetic , Histones , Chromatin/genetics , Enhancer Elements, Genetic/genetics , Gene Expression , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/genetics , Histones/metabolism
9.
Methods Mol Biol ; 2529: 327-403, 2022.
Article in English | MEDLINE | ID: mdl-35733023

ABSTRACT

Chemical modification of histone proteins by methylation plays a central role in chromatin regulation by recruiting epigenetic "readers" via specialized binding domains. Depending on the degree of methylation, the exact modified amino acid, and the associated reader proteins histone methylations are involved in the regulation of all DNA-based processes, such as transcription, DNA replication, and DNA repair. Here we present methods to identify histone methylation readers using a mass spectrometry-linked nucleosome affinity purification approach. We provide detailed protocols for the generation of semisynthetic methylated histones, their assembly into biotinylated nucleosomes, and the identification of methylation-specific nucleosome-interacting proteins from nuclear extracts via nucleosome pull-downs and label-free quantitative proteomics. Due to their versatility, these protocols allow the identification of readers of various histone methylations, and can also be adapted to different cell types and tissues, and other types of modifications.


Subject(s)
Histones , Nucleosomes , Chromatin , Histones/metabolism , Mass Spectrometry , Methylation , Protein Processing, Post-Translational
10.
Nucleic Acids Res ; 50(9): 5014-5028, 2022 05 20.
Article in English | MEDLINE | ID: mdl-35489065

ABSTRACT

The heterodimeric histone chaperone FACT, consisting of SSRP1 and SPT16, contributes to dynamic nucleosome rearrangements during various DNA-dependent processes including transcription. In search of post-translational modifications that may regulate the activity of FACT, SSRP1 and SPT16 were isolated from Arabidopsis cells and analysed by mass spectrometry. Four acetylated lysine residues could be mapped within the basic C-terminal region of SSRP1, while three phosphorylated serine/threonine residues were identified in the acidic C-terminal region of SPT16. Mutational analysis of the SSRP1 acetylation sites revealed only mild effects. However, phosphorylation of SPT16 that is catalysed by protein kinase CK2, modulates histone interactions. A non-phosphorylatable version of SPT16 displayed reduced histone binding and proved inactive in complementing the growth and developmental phenotypes of spt16 mutant plants. In plants expressing the non-phosphorylatable SPT16 version we detected at a subset of genes enrichment of histone H3 directly upstream of RNA polymerase II transcriptional start sites (TSSs) in a region that usually is nucleosome-depleted. This suggests that some genes require phosphorylation of the SPT16 acidic region for establishing the correct nucleosome occupancy at the TSS of active genes.


Subject(s)
Arabidopsis , Histone Chaperones , Nucleosomes , Transcription Initiation Site , Arabidopsis/genetics , Arabidopsis/metabolism , Chromatin/genetics , Histone Chaperones/metabolism , Histones/metabolism , Phosphorylation , RNA Polymerase II/metabolism , Transcriptional Elongation Factors/metabolism
11.
Mol Cell ; 81(14): 2944-2959.e10, 2021 07 15.
Article in English | MEDLINE | ID: mdl-34166609

ABSTRACT

A number of regulatory factors are recruited to chromatin by specialized RNAs. Whether RNA has a more general role in regulating the interaction of proteins with chromatin has not been determined. We used proteomics methods to measure the global impact of nascent RNA on chromatin in embryonic stem cells. Surprisingly, we found that nascent RNA primarily antagonized the interaction of chromatin modifiers and transcriptional regulators with chromatin. Transcriptional inhibition and RNA degradation induced recruitment of a set of transcriptional regulators, chromatin modifiers, nucleosome remodelers, and regulators of higher-order structure. RNA directly bound to factors, including BAF, NuRD, EHMT1, and INO80 and inhibited their interaction with nucleosomes. The transcriptional elongation factor P-TEFb directly bound pre-mRNA, and its recruitment to chromatin upon Pol II inhibition was regulated by the 7SK ribonucleoprotein complex. We postulate that by antagonizing the interaction of regulatory proteins with chromatin, nascent RNA links transcriptional output with chromatin composition.


Subject(s)
Chromatin/metabolism , RNA/metabolism , Transcription Factors/metabolism , Animals , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/metabolism , Gene Expression Regulation/physiology , HEK293 Cells , Humans , Male , Mice , Nucleosomes/metabolism , Positive Transcriptional Elongation Factor B/metabolism , Protein Binding/physiology , Proteomics/methods , RNA Polymerase II/metabolism , Transcription, Genetic/physiology , Transcriptional Elongation Factors/metabolism
12.
Nat Struct Mol Biol ; 26(12): 1184-1186, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31695189

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

13.
Nat Struct Mol Biol ; 26(10): 899-909, 2019 10.
Article in English | MEDLINE | ID: mdl-31548724

ABSTRACT

Polycomb repressive complex 2 (PRC2) maintains repression of cell-type-specific genes but also associates with genes ectopically in cancer. While it is currently unknown how PRC2 is removed from genes, such knowledge would be useful for the targeted reversal of deleterious PRC2 recruitment events. Here, we show that G-tract RNA specifically removes PRC2 from genes in human and mouse cells. PRC2 preferentially binds G tracts within nascent precursor mRNA (pre-mRNA), especially within predicted G-quadruplex structures. G-quadruplex RNA evicts the PRC2 catalytic core from the substrate nucleosome. In cells, PRC2 transfers from chromatin to pre-mRNA upon gene activation, and chromatin-associated G-tract RNA removes PRC2, leading to H3K27me3 depletion from genes. Targeting G-tract RNA to the tumor suppressor gene CDKN2A in malignant rhabdoid tumor cells reactivates the gene and induces senescence. These data support a model in which pre-mRNA evicts PRC2 during gene activation and provides the means to selectively remove PRC2 from specific genes.


Subject(s)
Polycomb Repressive Complex 2/metabolism , RNA Precursors/metabolism , Animals , Cell Line , Chromatin/metabolism , G-Quadruplexes , Histones/metabolism , Humans , Mice , Nucleosomes/metabolism , Protein Binding , RNA Precursors/chemistry , Transcriptional Activation
14.
Nat Struct Mol Biol ; 25(3): 225-232, 2018 03.
Article in English | MEDLINE | ID: mdl-29483650

ABSTRACT

Polycomb repressive complex 2 (PRC2) catalyzes methylation on lysine 27 of histone H3 (H3K27) and is required for maintaining transcriptional patterns and cellular identity, but the specification and maintenance of genomic PRC2 binding and H3K27 methylation patterns remain incompletely understood. Epigenetic mechanisms have been proposed, wherein pre-existing H3K27 methylation directs recruitment and regulates the catalytic activity of PRC2 to support its own maintenance. Here we investigate whether such mechanisms are required for specifying H3K27 methylation patterns in mouse embryonic stem cells (mESCs). Through re-expression of PRC2 subunits in PRC2-knockout cells that have lost all H3K27 methylation, we demonstrate that methylation patterns can be accurately established de novo. We find that regional methylation kinetics correlate with original methylation patterns even in their absence, and specification of the genomic PRC2 binding pattern is retained and specifically dependent on the PRC2 core subunit SUZ12. Thus, the H3K27 methylation patterns in mESCs are not dependent on self-autonomous epigenetic inheritance.


Subject(s)
Histones/metabolism , Polycomb Repressive Complex 2/metabolism , Animals , Cells, Cultured , CpG Islands , Embryonic Stem Cells/metabolism , Kinetics , Methylation , Mice
15.
Nucleic Acids Res ; 45(16): 9272-9289, 2017 Sep 19.
Article in English | MEDLINE | ID: mdl-28934504

ABSTRACT

Deposition of replication-independent histone variant H3.3 into chromatin is essential for many biological processes, including development and reproduction. Unlike replication-dependent H3.1/2 isoforms, H3.3 is expressed throughout the cell cycle and becomes enriched in postmitotic cells with age. However, lifelong dynamics of H3 variant replacement and the impact of this process on chromatin organization remain largely undefined. Using quantitative middle-down proteomics we demonstrate that H3.3 accumulates to near saturation levels in the chromatin of various mouse somatic tissues by late adulthood. Accumulation of H3.3 is associated with profound changes in global levels of both individual and combinatorial H3 methyl modifications. A subset of these modifications exhibit distinct relative abundances on H3 variants and remain stably enriched on H3.3 throughout the lifespan, suggesting a causal relationship between H3 variant replacement and age-dependent changes in H3 methylation. Furthermore, the H3.3 level is drastically reduced in human hepatocarcinoma cells as compared to nontumoral hepatocytes, suggesting the potential utility of the H3.3 relative abundance as a biomarker of abnormal cell proliferation activity. Overall, our study provides the first quantitative characterization of dynamic changes in H3 proteoforms throughout lifespan in mammals and suggests a role for H3 variant replacement in modulating H3 methylation landscape with age.


Subject(s)
Aging/metabolism , Histones/metabolism , Animals , Carcinoma, Hepatocellular/metabolism , Cells, Cultured , Hepatocytes/metabolism , Histone Code , Humans , Liver Neoplasms/metabolism , Lysine/metabolism , Male , Methylation , Mice, Inbred C57BL , Protein Isoforms/metabolism , Protein Processing, Post-Translational
16.
Nat Med ; 23(4): 483-492, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28263309

ABSTRACT

Diffuse intrinsic pontine glioma (DIPG) is an aggressive brain tumor that is located in the pons and primarily affects children. Nearly 80% of DIPGs harbor mutations in histone H3 genes, wherein lysine 27 is substituted with methionine (H3K27M). H3K27M has been shown to inhibit polycomb repressive complex 2 (PRC2), a multiprotein complex responsible for the methylation of H3 at lysine 27 (H3K27me), by binding to its catalytic subunit EZH2. Although DIPGs with the H3K27M mutation show global loss of H3K27me3, several genes retain H3K27me3. Here we describe a mouse model of DIPG in which H3K27M potentiates tumorigenesis. Using this model and primary patient-derived DIPG cell lines, we show that H3K27M-expressing tumors require PRC2 for proliferation. Furthermore, we demonstrate that small-molecule EZH2 inhibitors abolish tumor cell growth through a mechanism that is dependent on the induction of the tumor-suppressor protein p16INK4A. Genome-wide enrichment analyses show that the genes that retain H3K27me3 in H3K27M cells are strong polycomb targets. Furthermore, we find a highly significant overlap between genes that retain H3K27me3 in the DIPG mouse model and in human primary DIPGs expressing H3K27M. Taken together, these results show that residual PRC2 activity is required for the proliferation of H3K27M-expressing DIPGs, and that inhibition of EZH2 is a potential therapeutic strategy for the treatment of these tumors.


Subject(s)
Brain Stem Neoplasms/genetics , Cell Proliferation/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Glioma/genetics , Histones/genetics , Animals , Benzamides/pharmacology , Biphenyl Compounds , Brain Neoplasms/genetics , CRISPR-Cas Systems , Cell Line, Tumor , Chromatin Immunoprecipitation , Chromatography, Liquid , Cyclin-Dependent Kinase Inhibitor p16/drug effects , Cyclin-Dependent Kinase Inhibitor p16/genetics , Disease Models, Animal , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Gene Knockout Techniques , Glioblastoma/genetics , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Indazoles/pharmacology , Mice , Mice, SCID , Molecular Targeted Therapy , Morpholines , Mutation , Neoplasm Transplantation , Neural Stem Cells , Polycomb Repressive Complex 2/genetics , Pyridones/pharmacology , Tandem Mass Spectrometry , Tumor Suppressor Protein p14ARF/drug effects , Tumor Suppressor Protein p14ARF/genetics
17.
Mol Cell Proteomics ; 14(12): 3142-53, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26424599

ABSTRACT

Post-translational modifications (PTMs) of histone proteins play a fundamental role in regulation of DNA-templated processes. There is also growing evidence that proteolytic cleavage of histone N-terminal tails, known as histone clipping, influences nucleosome dynamics and functional properties. Using top-down and middle-down protein analysis by mass spectrometry, we report histone H2B and H3 N-terminal tail clipping in human hepatocytes and demonstrate a relationship between clipping and co-existing PTMs of histone H3. Histones H2B and H3 undergo proteolytic processing in primary human hepatocytes and the hepatocellular carcinoma cell line HepG2/C3A when grown in spheroid (3D) culture, but not in a flat (2D) culture. Using tandem mass spectrometry we localized four different clipping sites in H3 and one clipping site in H2B. We show that in spheroid culture clipped H3 proteoforms are mainly represented by canonical histone H3, whereas in primary hepatocytes over 90% of clipped H3 correspond to the histone variant H3.3. Comprehensive analysis of histone H3 modifications revealed a series of PTMs, including K14me1, K27me2/K27me3, and K36me1/me2, which are differentially abundant in clipped and intact H3. Analysis of co-existing PTMs revealed negative crosstalk between H3K36 methylation and H3K23 acetylation in clipped H3. Our data provide the first evidence of histone clipping in human hepatocytes and demonstrate that clipped H3 carry distinct co-existing PTMs different from those in intact H3.


Subject(s)
Hepatocytes/cytology , Histones/metabolism , Protein Processing, Post-Translational , Proteomics/methods , Acetylation , Cell Culture Techniques , Hep G2 Cells , Hepatocytes/metabolism , Histones/chemistry , Humans , Methylation , Tandem Mass Spectrometry/methods
18.
J Clin Endocrinol Metab ; 100(9): E1206-15, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26196948

ABSTRACT

CONTEXT: Current methods of preoperative diagnostics frequently fail to discriminate between benign and malignant thyroid neoplasms. In encapsulated follicular-patterned tumors (EnFPT), this discrimination is challenging even using histopathological analysis. Autoantibody response against tumor-associated antigens is a well-documented phenomenon with prominent diagnostic potential; however, autoantigenicity of thyroid tumors remains poorly explored. OBJECTIVES: Objectives were exploration of tumor-associated antigen repertoire of thyroid tumors and identification of candidate autoantibody biomarkers capable of discrimination between benign and malignant thyroid neoplasms. DESIGN, SETTING, AND PATIENTS: Proteins isolated from FTC-133 cells were subjected to two-dimensional Western blotting using pooled serum samples of patients originally diagnosed with either papillary thyroid carcinoma (PTC) or EnFPT represented by apparently benign follicular thyroid adenomas, as well as healthy individuals. Immunoreactive proteins were identified using liquid chromatography-tandem mass-spectrometry. Pathological reassessment of EnFPT was performed applying nonconservative criteria for capsular invasion and significance of focal PTC nuclear changes (PTC-NCs). Recombinant T-complex protein 1 subunitζ (TCP-1ζ) was used to examine an expanded serum sample set of patients with various thyroid neoplasms (n = 89) for TCP-1ζ autoantibodies. All patients were included in tertiary referral centers. RESULTS: A protein demonstrating a distinct pattern of EnFPT-specific seroreactivity was identified as TCP-1ζ protein. A subsequent search for clinicopathological correlates of TCP-1ζ seroreactivity revealed nonclassical capsular invasion or focal PTC-NC in all TCP-1ζ antibody-positive cases. Further studies in an expanded sample set confirmed the specificity of TCP-1ζ autoantibodies to malignant EnFPT. CONCLUSIONS: We identified TCP-1ζ autoantibodies as a potential biomarker for presurgical discrimination between benign and malignant encapsulated follicular-patterned thyroid tumors. Our results suggest the use of nonconservative morphological criteria for diagnosis of malignant EnFPT in biomarker identification studies and provide a peculiar example of uncovering the diagnostic potential of a candidate biomarker using incorporation of pathological reassessment in the pipeline of immunoproteomic research.


Subject(s)
Adenocarcinoma, Follicular/diagnosis , Autoantibodies/blood , Carcinoma, Papillary/diagnosis , Chaperonin Containing TCP-1/immunology , Thyroid Neoplasms/diagnosis , Adenocarcinoma, Follicular/blood , Adenocarcinoma, Follicular/immunology , Adenocarcinoma, Follicular/pathology , Adult , Biomarkers/blood , Carcinoma, Papillary/blood , Carcinoma, Papillary/immunology , Carcinoma, Papillary/pathology , Female , Humans , Middle Aged , Thyroid Neoplasms/blood , Thyroid Neoplasms/immunology , Thyroid Neoplasms/pathology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...